Estradiol metabolites as isoform-specific inhibitors of human glutathione S-transferases

https://doi.org/10.1016/j.cbi.2004.10.006Get rights and content

Abstract

Numerous studies have suggested that the lifetime dose of unopposed estrogen is a significant risk factor for breast and uterine cancer. Estradiol (E2) plays a putative role as a tumor promoter through interaction with estrogen receptors but can also be metabolized to redox active and/or mutagenic semiquinones and quinones. Similarly, equine estrogens (components of certain hormone replacement therapy preparations) are converted to quinone metabolites. The use of hormone replacement therapy has also been associated with increased breast and endometrial cancer risk. Recently, metabolites of certain equine estrogens have been shown to inhibit human glutathione S-transferases (hGSTs). Since E2 and equine estrogens share similarities in other biological interactions, we have investigated the inhibitory capacity of endogenously formed E2 metabolites toward various hGSTs. The quinone metabolite of 2-hydroxy-17-β-estradiol (2-OH-E2) was synthesized, and inhibition of hGST-mediated biotransformation of model substrates was assessed. Inhibition of purified recombinant hGSTM1-1 and hGSTA1-1 occurred in a concentration-dependent manner with IC50-values of approximately 250 and 350 nM, respectively. hGSTs M2-2, P1-1 and T1-1 were significantly less sensitive to inhibition. Specific glutathione-conjugates of the estrogen quinone also potently inhibited hGSTM1-1 and hGSTA1-1. Mass spectrometry data indicate that the inhibition was not mediated via covalent adduction. Although we have demonstrated hGST inhibition via E2 metabolites, our findings indicate that the isoform specificity and potency of GST inhibition by endogenous E2 metabolites is different than that of equine estrogen metabolites.

Introduction

The glutathione S-transferases (GSTs) are a multigene family of biotransformation enzymes capable of catalyzing glutathione (GSH) conjugation to exogenous and endogenous electrophilic species (for review see [1]). The GSTs have also been implicated in modulation of stress-activated protein kinase activity [2], [3], [4], protection against lipid peroxidation [5], [6] and transport of cellular molecules such as steroids [7]. The mammalian GST family is divided into at least eight classes (alpha (A), kappa (K), mu (M), omega (O), pi (P), sigma (S), theta (T), and zeta (Z)) with multiple subfamilies per class [8], [9], [10], [11], [12], [13], [14]. Individual GST isoforms demonstrate unique, though often overlapping, substrate specificity and range of cellular functions [1].

Estradiol (E2) is a major circulating form of estrogen in humans and has been associated with the development of breast and endometrial cancers (for review see [15], [16]). E2 is thought to increase cell proliferation via estrogen receptor-mediated cell signaling thereby acting as a tumor promoter; however, oxidative metabolites of E2 are also considered to be mutagens (for review see [17]). To combat symptoms of menopause associated in part with declining endogenous estrogen levels, hormone replacement therapy is often prescribed. However, this treatment is similarly associated with increased risk of estrogen-related cancers [18], [19].

Equine estrogens (i.e. equilin and equilenin) are major components of certain estrogen replacement therapies such as Premarin (Wyeth-Ayrest) [20]. E2, equilin and equilenin are metabolized to catechol estrogens and further to quinones via autooxidation (in the cases of equilin and equilenin) or via cytochrome P450 and/or peroxidase mediated catalysis (in the case of E2) (Fig. 1) [20], [21], [22], [23], [24], [25], [26]. Upon quinone formation, the molecules may spontaneously react with glutathione [27], [28].

Metabolism is thought to modulate estrogen toxicity. For instance, quinone metabolites are capable of DNA adduction and free radical generation [29], [30], and in vivo research indicates that 4-hydroxylation is an activation reaction [31]. In the Syrian hamster and CD1 mouse models, 1,3,5(10)-estratriene-3,4,17β-triol (4-OH-E2) treatment results in higher tumor incidence than 1,3,5(10)-estratriene-2,3,17β-triol (2-OH-E2) treatment [31], [32]. Furthermore, in target organs sensitive to estrogen-induced cancer, 4-hydroxylation of estradiol predominates over 2-hydroxylation [33], and as opposed to the stable DNA adducts associated with E22,3-Q, E23,4-Q forms depurinating adducts with DNA [34], [35].

It has recently been established that metabolites of equine estrogens, equilenin and equilin, specifically and irreversibly inhibit human GSTs (hGSTs) [36], [37], [38]. Inhibition of hGSTM1-1 and hGSTP1-1 occurred at concentrations in the micromolar range via irreversible adduction to cysteine residues and oxidative damage [36], [37]. However, whether hGST inhibition by equine estrogens is unique in potency and isoform specificity, or whether endogenously formed estradiol metabolites are capable of similar inhibitory effects on human GSTs is unknown. Therefore, we sought to determine whether reactive metabolites of E2 have interactions with GST similar in isoform specificity and potency to exogenously administered equine estrogens. Our aim was to characterize GST inhibition by endogenous quinone and glutathione conjugate metabolites of E2. We have identified isoform-specific inhibition of hGSTs with IC50values in the nanomolar range.

Section snippets

Abbreviations

2-OH-E2, 1,3,5(10)-estratriene-2,3,17β-triol; 4-OH-E2, 1,3,5(10)-estratriene-3,4,17β-triol; E12,3-Q, 1(10),4(5)-estradiene-2,3,17-trione; E22,3-Q, 17β-hydroxy-1(10),4(5)-estradiene-2,3-dione; E23,4-Q, 17β-hydroxy-1,5(10)-estradiene-3,4-dione; 2-OH-1-GSyl-E2, 2-hydroxy-1-glutathion-S-yl-17β-estradiol; 2-OH-4-GSyl-E2, 2-hydroxy-4-glutathion-S-yl-17β-estradiol; 4-OH-2-GSyl-E2, 4-hydroxy-2-glutathion-S-yl-17β-estradiol. As previously suggested [27], the single monoglutathione conjugate formed upon

hGST inhibition: E22,3-Q

To determine whether E2 metabolites are inhibitors of hGST isoforms (GSTs A1-1, M1-1, M2-2, P1-1, T1-1), we examined hGST-mediated biotransformation of model substrates in the presence and absence of E22,3-Q. E22,3-Q exhibited potent inhibition in assays of hGSTM1-1 and hGSTA1-1 activities toward CDNB (Fig. 2). This inhibition was concentration dependent, demonstrating IC50-values of approximately 250 and 350 nM for hGSTM1-1 and hGSTA1-1, respectively. Interestingly, E22,3-Q preincubation with

Discussion

Quinones and quinone–thioethers are known to inhibit GSTs [48], [49]. For instance, covalent and irreversible GST inhibition by halogenated quinones is most effective when a glutathione moiety is added [49]. The resulting quinone–thioether is both reactive toward thiol groups and targeted to GST glutathione-binding active site. Our results generated with glutathione-conjugated metabolites of E2 do not conform to this paradigm. Although glutathione conjugation may confer inhibitory potential, no

Acknowledgements

The authors wish to acknowledge Portia Vliet for her excellent technical assistance in purifying the histidine-tagged hGSTT1-1. This work was supported in part by NIEHS Center Grant P30-ES07033, R01 ES05780, R01 AG17635 and NIA Training Grant T32 AG00057.

References (58)

  • S. Raghunathan et al.

    Crystal structure of human class mu glutathione transferase GSTM2-2. Effects of lattice packing on conformational heterogeneity

    J. Mol. Biol.

    (1994)
  • V. Cortes-Gallegos et al.

    Estrogen peripheral levels vs. estrogen tissue concentration in the human female reproductive tract

    J. Steroid Biochem.

    (1975)
  • J.G. Bisseling et al.

    Glutathione S-transferases in human ovarian follicular fluid

    Fertil. Steril.

    (1997)
  • D.L. Eaton et al.

    Concise review of the glutathione S-transferases and their significance to toxicology

    Toxicol. Sci.

    (1999)
  • V. Adler et al.

    Regulation of JNK signaling by GSTp

    Embo. J.

    (1999)
  • Z. Yin et al.

    Glutathione S-transferase p elicits protection against H2O2-induced cell death via coordinated regulation of stress kinases

    Cancer Res.

    (2000)
  • Y. Yang et al.

    Protection of HLE B-3 cells against hydrogen peroxide- and naphthalene-induced lipid peroxidation and apoptosis by transfection with hGSTA1 and hGSTA2

    Invest. Ophthalmol. Vis. Sci.

    (2002)
  • G. Litwack et al.

    Ligandin: a hepatic protein, which binds steroids, bilirubin, carcinogens and a number of exogenous organic anions

    Nature

    (1971)
  • P.G. Board et al.

    Identification of novel glutathione transferases and polymorphic variants by expressed sequence tag database analysis

    Drug Metab. Dispos.

    (2001)
  • J.M. Harris et al.

    A novel glutathione transferase (13-13) isolated from the matrix of rat liver mitochondria having structural similarity to class theta enzymes

    Biochem. J.

    (1991)
  • B. Mannervik et al.

    Identification of three classes of cytosolic glutathione transferase common to several mammalian species: correlation between structural data and enzymatic properties

    Proc. Natl. Acad. Sci. U.S.A.

    (1985)
  • D.J. Meyer et al.

    Theta a new class of glutathione transferases purified from rat and man

    Biochem. J.

    (1991)
  • D.J. Meyer et al.

    Characterization of rat spleen prostaglandin H D-isomerase as a sigma-class GSH transferase

    Biochem. J.

    (1995)
  • S.E. Pemble et al.

    Glutathione S-transferase class Kappa: characterization by the cloning of rat mitochondrial GST and identification of a human homologue

    Biochem. J.

    (1996)
  • B.E. Henderson et al.

    Hormonal carcinogenesis

    Carcinogenesis

    (2000)
  • R. Kaaks et al.

    Obesity endogenous hormones, and endometrial cancer risk: a synthetic review

    Cancer Epidemiol. Biomarkers Prev.

    (2002)
  • J.D. Yager

    Endogenous estrogens as carcinogens through metabolic activation

    J. Natl. Cancer Inst. Monogr.

    (2000)
  • F. Zhang et al.

    The major metabolite of equilin, 4-hydroxyequilin, autoxidizes to an o-quinone which isomerizes to the potent cytotoxin 4-hydroxyequilenin-o-quinone

    Chem. Res. Toxicol.

    (1999)
  • K. Cao et al.

    Covalent binding of catechol estrogens to glutathione catalyzed by horseradish peroxidase, lactoperoxidase, or rat liver microsomes

    Chem. Res. Toxicol.

    (1998)
  • Cited by (19)

    • Gut microbial β-glucuronidases reactivate estrogens as components of the estrobolome that reactivate estrogens

      2019, Journal of Biological Chemistry
      Citation Excerpt :

      As such, steroid hormone biosynthetic pathways have been under investigation for decades, and there is abundant information on their metabolism in humans. Cytochromes P450, UGTs, sulfotransferases, and catechol-O-methyltransferases are just a few of the major enzymatic families that metabolize estrone, for example (37–41). Some of the major metabolites of estrone are highlighted in Fig. 7.

    • Metabolism of Glutathione S-Conjugates: Multiple Pathways

      2018, Comprehensive Toxicology: Third Edition
    • Glutathione transferase polymorphisms and risk of endometriosis associated with polychlorinated biphenyls exposure in Italian women: A gene-environment interaction

      2012, Fertility and Sterility
      Citation Excerpt :

      To evaluate the possibility that, apart from GSTM1 and GSTT1, other GST enzymes could have a role in the development of endometriosis, we analyzed two other polymorphisms at GSTA1 and GSTP1 genes, which, lowering tissue defenses against ROS, may also influence disease susceptibility. To our knowledge no other study analyzed a possible association between GSTA1 C-69T polymorphism and endometriosis; nevertheless it could be functionally relevant, being expressed both in the ovaries and in the uterus (43). Previous data about the association of GSTP1 and endometriosis were inconclusive.

    • Enzymes Involved in Processing Glutathione Conjugates

      2010, Comprehensive Toxicology, Second Edition
    View all citing articles on Scopus
    1

    Present address: University of Texas MD Anderson Cancer Center, Department of Carcinogenesis, Smithvile, TX 78957, USA.

    View full text